1
|
Reglero-Real N, Rolas L, Nourshargh S. Aging microvasculature: Effects on immune cell trafficking and inflammatory diseases. J Exp Med 2025; 222:e20242154. [PMID: 40455014 PMCID: PMC12128883 DOI: 10.1084/jem.20242154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 06/11/2025] Open
Abstract
Leukocyte recruitment to sites of inflammation is vital for orchestrating an effective immune response. Key to this process is the ability of leukocytes to migrate through venular walls, engaging in sequential interactions with endothelial cells, pericytes, and the venular basement membrane. The aging process exerts profound effects on the molecular and functional properties of the vasculature, thereby influencing the profile and dynamics of leukocyte trafficking during inflammation. In this review, by focusing mainly on neutrophils, we summarize key examples of how the aged microvasculature and perivascular stroma cells promote dysregulated leukocyte-venular wall interactions and present the associated molecular mechanisms. Additionally, we discuss the functional implications of such aberrant leukocyte behavior to age-related and chronic inflammatory pathologies.
Collapse
Affiliation(s)
- Natalia Reglero-Real
- Departamento de Biología Molecular, Instituto Universitario de Biología Molecular (IUBM) and Centro de Biología Molecular Severo Ochoa (CBM), Universidad Autónoma de Madrid, UAM-CSIC, Madrid, Spain
- Instituto de Investigación Sanitaria La Princesa, Madrid, Spain
| | - Loïc Rolas
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- Centre for Microvascular Research, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
2
|
Wang Y, Liu Z, Hu H, Cheng W, Chen Z, Zhong J, Dai J, Zhang J. Porcine urinary bladder matrix encapsulated biological patch enhances dural repair and promotes anti-adhesion via mesothelial cell induction. Colloids Surf B Biointerfaces 2025; 254:114801. [PMID: 40412291 DOI: 10.1016/j.colsurfb.2025.114801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/25/2025] [Accepted: 05/12/2025] [Indexed: 05/27/2025]
Abstract
Decellularized biomaterial-based dural patches are clinically employed as dura mater substitutes, owing to their biocompatibility, biodegradability, mechanical flexibility, and efficacy in promoting tissue regeneration. However, these biological grafts have challenges related to patch durability, cerebrospinal fluid leakage, and brain tissue adhesion. In this study, we developed a composite dural patch with a biomimetic structure by encapsulating multilayered small intestinal submucosa (SIS) membranes using basement membrane-containing urinary bladder matrix (UBM) membranes as outer layers, to enhance dural repair. The newly designed UBM@SIS patch demonstrates superior mechanical properties, excellent tissue regeneration capabilities, and enhanced anti-adhesion effects. Immunofluorescent staining revealed a densely organized cell layer anchored to the surface of the UBM@SIS patch, identified as mesothelial cells (Pan-Cytokeratin+/E-cadherin+) and arachnoid barrier cells (ABCs), which are essential for lubricating brain tissue, preventing adhesion, and reducing cerebrospinal fluid leakage. In contrast, only a sparse presence of Pan-Cytokeratin-labeled mesothelial cells and E-cadherin-labeled ABCs was observed on the SIS patch, exhibiting incomplete pia mater and significant adhesion on interface between patch and brain tissue. Furthermore, neurotoxicity evaluations confirmed that the encapsulation of UBM reduces astrogliosis and neuroinflammatory response in early stage. These results suggest that the novel UBM@SIS patch effectively addresses the limitations of existing biological patches and holds significant potential for clinical application.
Collapse
Affiliation(s)
- Yulu Wang
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Zhentao Liu
- Department of Neurosurgery, No.988 Hospital of Joint Logistic Support Force, Henan Province 454000, China
| | - Hongkang Hu
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Wenyue Cheng
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Zhaoxin Chen
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jie Zhong
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Jing Dai
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jian Zhang
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
3
|
Zeng J, Wang J, Zhang Y, Wang Z, Zhu Y, Hou Y, Li X, Peng H, Lobie PE, Ma S. Mesenchymal stem cells attenuate diabetic vascular complication by reducing irregular extracellular matrix production in human blood vessel organoids. LAB ON A CHIP 2025. [PMID: 40341804 DOI: 10.1039/d5lc00107b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Mesenchymal stem cells (MSCs) hold potential for treating diabetic vascular complications, but current models fail to adequately replicate the complexities of diabetic vascular disease, limiting our ability to accurately assess their therapeutic effects. To this end, we developed a co-culture system using a combination of human embryonic stem cell-derived blood vessel organoids (BVOs) and MSCs. This system could accurately replicate key aspects of diabetic pathology, including basement membrane thickening and excessive extracellular matrix (ECM) deposition. The results showed that MSCs were effective in attenuating basement membrane thickening and reducing ECM deposition in BVOs under diabetic conditions. Subsequent transcriptomics demonstrated that the MSC-treated group exhibited a notable normalization of ECM-related gene expression, particularly in collagen IV levels. Furthermore, the inhibition of the NF-κB signaling pathway was identified as a crucial mechanism underlying the therapeutic efficacy of MSCs. This study demonstrates the potential of MSCs to counteract diabetic vascular complications and emphasizes the co-culture system as a more physiologically relevant model to investigate the preventive and therapeutic potential of MSCs in diabetic pathology.
Collapse
Affiliation(s)
- Junhong Zeng
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Jiaqi Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Yu Zhang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
- Key Lab of Industrial Biocatalysis Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yu Zhu
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Yibo Hou
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Xiangsai Li
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
| | - Haiying Peng
- General Hospital of the Southern Theater Command of the Chinese People's Liberation Army, Guangzhou 510010, China
| | - Peter E Lobie
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
- Key Lab of Industrial Biocatalysis Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China.
- Key Lab of Industrial Biocatalysis Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
4
|
Zeng J, Heilig S, Ryma M, Groll J, Li C, Matsusaki M. Outermost Cationic Surface Charge of Layer-by-Layer Films Prevents Endothelial Cells Migration for Cell Compartmentalization in Three-Dimensional Tissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2417538. [PMID: 39985273 PMCID: PMC12097075 DOI: 10.1002/advs.202417538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Indexed: 02/24/2025]
Abstract
Tissues and organs possess an organized cellular arrangement that enables their unique functions. However, conventional three-dimensional (3D) encapsulation techniques fail to recapitulate this complexity due to the cell migration during cell culture. In biological tissues, basement membranes (BMs) are essential to mechanically support cellular organization. This study finds that a positively charged outermost surface of multilayered nanofilms, fabricated through layer-by-layer assembly of poly-l-lysine (PLL) and dextran (Dex) via hydrogen bonds, stimulates the barrier functions of BMs. This type of artificial BM (A-BM) demonstrates enhanced barrier properties in comparison to other types of A-BMs composed of BM components such as collagen type IV and laminin. Such an enhancement is potentially associated with the outermost cationic layer, which inhibits the sprouting of endothelial cells (ECs) and effectively prevents EC migration over a 14-d period, aligning with the formation timeline of natural BMs in 3D tissues. Finally, 3D organized vascular channels are successfully engineered with the support of shape-adaptable PLL/Dex nanofilms. This approach offers a guideline for engineering organized 3D tissue models by regulating cell migration, which can provide reliable platforms for in vitro permeability assay of new drugs or drug delivery carriers.
Collapse
Affiliation(s)
- Jinfeng Zeng
- College of TextilesDonghua UniversityShanghai201620China
- Department of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
| | - Sven Heilig
- University of WürzburgPleicherwall 297070WürzburgGermany
| | - Matthias Ryma
- University of WürzburgPleicherwall 297070WürzburgGermany
| | - Jürgen Groll
- University of WürzburgPleicherwall 297070WürzburgGermany
| | - Congju Li
- College of TextilesDonghua UniversityShanghai201620China
| | - Michiya Matsusaki
- Department of Applied ChemistryGraduate School of EngineeringOsaka University2‐1 YamadaokaSuitaOsaka565–0871Japan
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory ChemistryOsaka UniversitySuitaOsakaJapan
| |
Collapse
|
5
|
Zhao N, Pessell AF, Chung TD, Searson PC. Brain vascular basement membrane: comparison of human and mouse brain at the transcriptomic and proteomic levels. Matrix Biol 2025:S0945-053X(25)00036-8. [PMID: 40294830 DOI: 10.1016/j.matbio.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
The cerebrovascular basement membrane (BM) is a key component of the blood-brain barrier (BBB). The BM provides structural support for brain microvascular endothelial cells and the supporting cells of the neurovascular unit, and facilitates cell signaling through adhesion receptors, regulates the concentration of soluble factors, and serves as an additional barrier for transport. However, our understanding of the composition of BM remains incomplete. Here we analyze recent proteomic and genomic data to assess the composition of BM in human and mouse brain, and in tissue-engineered BBB models. All data sets confirm that the main components of brain BM are collagen IV a1/2, laminin, along with agrin, perlecan, and nidogen. Transcriptomic data from human BMECs suggests that the main laminin isoform is Laminin 321, while transcriptomic data from mice and proteomic data from mice and humans suggest that Laminin 521 is the predominant isoform. Transcriptomic data from iBMECs suggest that Laminin 511 is the predominant isoform. The supporting molecules agrin, perlecan, and nidogen were detected at significant levels in all studies, although only nidogen 1 was detected in the human transcriptomic data sets. No significant differences in human BM composition were observed in BMECs along the arterio-venous axis, or in comparison of healthy and AD brains.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tracy D Chung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
6
|
Brunmaier LAE, Ozdemir T, Walker TW. Angiogenesis: Biological Mechanisms and In Vitro Models. Ann Biomed Eng 2025:10.1007/s10439-025-03721-2. [PMID: 40210793 DOI: 10.1007/s10439-025-03721-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/12/2025]
Abstract
The translation of biomedical devices and drug research is an expensive and long process with a low probability of receiving FDA approval. Developing physiologically relevant in vitro models with human cells offers a solution to not only improving the odds of FDA approval but also to expand our ability to study complex in vivo systems in a simpler fashion. Animal models remain the standard for pre-clinical testing; however, the data from animal models is an unreliable extrapolation when anticipating a human response in clinical trials, thus contributing to the low rates of translation. In this review, we focus on in vitro vascular or angiogenic models because of the incremental role that the vascular system plays in the translation of biomedical research. The first section of this review discusses the most common angiogenic cytokines that are used in vitro to initiate angiogenesis, followed by angiogenic inhibitors where both initiators and inhibitors work to maintain vascular homeostasis. Next, we evaluate previously published in vitro models, where we evaluate capturing the physical environment for biomimetic in vitro modeling. These topics provide a foundation of parameters that must be considered to improve and achieve vascular biomimicry. Finally, we summarize these topics to suggest a path forward with the goal of engineering human in vitro models that emulate the in vivo environment and provide a platform for biomedical device and drug screening that produces data to support clinical translation.
Collapse
Affiliation(s)
- Laura A E Brunmaier
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Tugba Ozdemir
- Nanoscience and Biomedical Engineering Department, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA
| | - Travis W Walker
- Karen M. Swindler Department of Chemical and Biological Engineering, South Dakota School of Mines & Technology, 501 E St. Joseph St., Rapid City, SD, 57701, USA.
| |
Collapse
|
7
|
Leclech C, Cardillo G, Roellinger B, Zhang X, Frederick J, Mamchaoui K, Coirault C, Barakat AI. Micro-Scale Topography Triggers Dynamic 3D Nuclear Deformations. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410052. [PMID: 39873289 PMCID: PMC11923911 DOI: 10.1002/advs.202410052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/29/2024] [Indexed: 01/30/2025]
Abstract
Navigating complex extracellular environments requires extensive deformation of cells and their nuclei. Most in vitro systems used to study nuclear deformations impose whole-cell confinement that mimics the physical crowding experienced by cells during 3D migration through tissues. Such systems, however, do not reproduce the types of nuclear deformations expected to occur in cells that line tissues such as endothelial or epithelial cells whose physical confinement stems principally from the topography of their underlying basement membrane. Here, it is shown that endothelial cells and myoblasts cultured on microgroove substrates that mimic the anisotropic topography of the basement membrane exhibit large-scale 3D nuclear deformations, with partial to complete nuclear penetration into the microgrooves. These deformations do not lead to significant DNA damage and are dynamic with nuclei cyclically entering and exiting the microgrooves. Atomic force microscopy measurements show that these deformation cycles are accompanied by transient changes in perinuclear stiffness. Interestingly, nuclear penetration into the grooves is driven principally by cell-substrate adhesion stresses, with a limited need for cytoskeleton-associated forces. Finally, it is demonstrated that myoblasts from laminopathy patients exhibit abnormal nuclear deformations on microgrooves, raising the possibility of using microgroove substrates as a novel functional diagnostic platform for pathologies that involve abnormal nuclear mechanics.
Collapse
Affiliation(s)
- Claire Leclech
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| | - Giulia Cardillo
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| | - Bettina Roellinger
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| | - Xingjian Zhang
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| | - Joni Frederick
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| | - Kamel Mamchaoui
- Sorbonne UniversitéINSERM UMRS‐974Centre for Research in Myology GH Pitié‐Salpêtrière 47 bd de l'HôpitalParis75013France
| | - Catherine Coirault
- Sorbonne UniversitéINSERM UMRS‐974Centre for Research in Myology GH Pitié‐Salpêtrière 47 bd de l'HôpitalParis75013France
| | - Abdul I. Barakat
- LadHyXCNRSEcole PolytechniqueInstitut Polytechnique de ParisPalaiseau91120France
| |
Collapse
|
8
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2025; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
9
|
Lee Y, Tukei KL, Fang Y, Kuila S, Liu X, Imoukhuede PI. Integrative analysis of angiogenic signaling in obesity: capillary features and VEGF binding kinetics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.23.630107. [PMID: 39763822 PMCID: PMC11703262 DOI: 10.1101/2024.12.23.630107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Obesity is a global health crisis, with its prevalence particularly severe in the United States, where over 42% of adults are classified as obese. Obesity is driven by complex molecular and tissue-level mechanisms that remain poorly understood. Among these, angiogenesis-primarily mediated by vascular endothelial growth factor (VEGF-A)-is critical for adipose tissue expansion but presents unique challenges for therapeutic targeting due to its intricate regulation. Systems biology approaches have advanced our understanding of VEGF-A signaling in vascular diseases, but their application to obesity is limited by scattered and sometimes contradictory data. To address this gap, we performed a comprehensive analysis of the existing literature to synthesize key findings, standardize data, and provide a holistic perspective on the adipose vascular microenvironment. The data mining revealed five key findings: (1) obesity increases adipocyte size by 78%; (2) vessel density in adipose tissue decreases by 51% in obese mice, with vessels being 47-58% smaller and 4-9 times denser in comparison with tumor vessels; (3) capillary basement membrane thickness remains similar regardless of obesity; (4) VEGF-A shows the strongest binding affinity for VEGFR1, with four times stronger affinity for VEGFR2 than for NRP1; and (5) binding affinities measured by radioligand binding assay and surface plasmon resonance (SPR) are significantly different. These consolidated findings provide essential parameters for systems biology modeling, new insights into obesity-induced changes in adipose tissue, and a foundation for developing angiogenesis-targeting therapies for obesity.
Collapse
|
10
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
11
|
Zhong W, Chen J, Xie Q, Cheng W, Zhao M, Sun Y, Dai J, Zhang J. A Novel UBM/SIS Composite Biological Scaffold for 2-Year Abdominal Defect Repairing and Strength Recovery in Canine Model. Adv Biol (Weinh) 2025; 9:e2400131. [PMID: 39542874 DOI: 10.1002/adbi.202400131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/21/2024] [Indexed: 11/17/2024]
Abstract
Biological scaffolds are widely utilized in hernia treatment due to their exceptional pro-regenerative properties, which mitigate scar formation. However, serious complications occurred, caused by inflammatory response, premature degradation, and mechanical failure. Consequently, improvements of the biological scaffold are necessary to mitigate these risks. In this study, a novel biological scaffold integrating basement membrane-containing urinary bladder matrix (UBM) and small intestinal submucosa (SIS) is developed, and its safety and effectiveness are assessed in comparison to a commercial SIS (c-SIS) scaffold. The introduction of UBM as top surface layers significantly promotes cell adhesion, facilitating rapid formation of isolated regeneration zone. Proteomic analysis has demonstrated a more efficient decellularization of the UBM/SIS scaffold, which subsequently mitigates inflammation in murine models, and promotes the polarization of macrophages toward the pro-healing M2 phenotype in a rat model of abdominal wall muscle defect. Furthermore, a two-year repair trial is conducted on a full-thickness abdominal wall muscle defect in canine model and confirmed that the UBM/SIS scaffold exhibits reduced seroma occurrences and enhanced tissue repair performances. Overall, the efficacy of this novel biological scaffold suggests its potential to minimize hernia recurrence in clinical practice and mitigate patient suffering from severe inflammatory responses.
Collapse
Affiliation(s)
- Weidong Zhong
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
- Department of Gastrointestinal Surgery, Jiangyin People's Hospital Affiliated to Nantong University, Jiangyin, 214400, China
| | - Jinshui Chen
- Department of General Surgery, The 991st Hospital of Joint Logistic Support Force of People's Liberation Army, Xiangyang, Hubei, 441003, China
| | - Qifeng Xie
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Wenyue Cheng
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Meibiao Zhao
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Yang Sun
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jing Dai
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| | - Jian Zhang
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai, 200003, China
| |
Collapse
|
12
|
Hansen CE, Hollaus D, Kamermans A, de Vries HE. Tension at the gate: sensing mechanical forces at the blood-brain barrier in health and disease. J Neuroinflammation 2024; 21:325. [PMID: 39696463 PMCID: PMC11657007 DOI: 10.1186/s12974-024-03321-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024] Open
Abstract
Microvascular brain endothelial cells tightly limit the entry of blood components and peripheral cells into the brain by forming the blood-brain barrier (BBB). The BBB is regulated by a cascade of mechanical and chemical signals including shear stress and elasticity of the adjacent endothelial basement membrane (BM). During physiological aging, but especially in neurological diseases including multiple sclerosis (MS), stroke, small vessel disease, and Alzheimer's disease (AD), the BBB is exposed to inflammation, rigidity changes of the BM, and disturbed cerebral blood flow (CBF). These altered forces lead to increased vascular permeability, reduced endothelial reactivity to vasoactive mediators, and promote leukocyte transmigration. Whereas the molecular players involved in leukocyte infiltration have been described in detail, the importance of mechanical signalling throughout this process has only recently been recognized. Here, we review relevant features of mechanical forces acting on the BBB under healthy and pathological conditions, as well as the endothelial mechanosensory elements detecting and responding to altered forces. We demonstrate the underlying complexity by focussing on the family of transient receptor potential (TRP) ion channels. A better understanding of these processes will provide insights into the pathogenesis of several neurological disorders and new potential leads for treatment.
Collapse
Affiliation(s)
- Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - David Hollaus
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
| | - Alwin Kamermans
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands.
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
13
|
Saijo Y, Ichinose S, Dohi T, Ogawa R. Vascular Basement Membrane Fragmentation in Keloids and the Expression of Key Basement Membrane Component Genes. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e6366. [PMID: 39717721 PMCID: PMC11666161 DOI: 10.1097/gox.0000000000006366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 10/09/2024] [Indexed: 12/25/2024]
Abstract
Background Keloids are growing scars that arise from injury to the reticular dermis and subsequent chronic local inflammation. The latter may be promoted by vascular hyperpermeability, which permits the ingress of chronic inflammatory cells/factors. Cutaneous capillaries consist of endothelial cells that generate, and are anchored by, a vascular basement membrane (VBM). Because VBM blocks immune cells/factors ingress, we investigated whether keloids are associated with altered VBM structure and/or VBM component expression by local endothelial cells. Methods In total, 54 keloid (n = 27) and adjacent normal skin (n = 27) samples from 14 patients underwent transmission electron microscopy (TEM). Cross-sections of whole capillaries were identified. VBM thickness, continuity, and the number of layers in keloid and normal skin tissues were quantified. The differential expression of 222 previously reported VBM component genes in keloid and normal skin endothelial cells was analyzed using the GSE121618-microarray dataset. Results TEM images showed that keloid VBMs were significantly thinner than adjacent skin VBMs (0.053 versus 0.078 nm; P < 0.001). They were also greatly fragmented (continuity was 46% versus 85% in normal skin; P < 0.001) and had fewer (1.2 versus 2.4) layers (P < 0.001). Keloidal endothelial cells demonstrated downregulation of 22 genes, including papilin, laminin-α5, and laminin-α2, and upregulation of 28 genes, including laminin-β1, laminin-β2, laminin-γ1, and laminin-γ2. Conclusions VBMs are greatly fragmented in keloids. These changes support the notion that keloids are initiated/promoted, at least partly, by vascular hyperpermeability.
Collapse
Affiliation(s)
- Yusaku Saijo
- From the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School Hospital, Tokyo, Japan
| | - Shizuko Ichinose
- From the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School Hospital, Tokyo, Japan
| | - Teruyuki Dohi
- From the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School Hospital, Tokyo, Japan
| | - Rei Ogawa
- From the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School Hospital, Tokyo, Japan
| |
Collapse
|
14
|
Resnikoff HA, Schwarzbauer JE. Increased basal fibronectin is sufficient to promote excess endothelial cell matrix assembly causing localized barrier dysfunction. Mol Biol Cell 2024; 35:ar120. [PMID: 39046775 PMCID: PMC11449387 DOI: 10.1091/mbc.e24-02-0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Endothelial cell behavior is regulated by subendothelial extracellular matrix (ECM). The ECM protein fibronectin (FN) is rare in healthy blood vessels but accumulates in disease accompanied by endothelial dysfunctions. Here, we report that excess assembly of FN disrupts key endothelial functions. We mimicked increased FN expression as in diseased stroma by providing exogenous FN basally in a Transwell insert and found dose-dependent upregulation of subendothelial FN matrix assembly. Taking advantage of discontinuous matrix assembly by endothelial cells, we show correlations between regional increases in FN matrix and disruptions in endothelial cell morphology, VE-cadherin junctions, and the cell cycle, all of which were not changed in FN-deficient regions of the monolayer. These changes affected endothelial barrier function with increased monolayer permeability exposing basal regions of high FN matrix and permitting FN-dependent adhesion of MDA-MB-231 tumor cells from the apical side of the monolayer. FN matrix accumulation was quick and increases in FN matrix preceded all other changes in the endothelium. Therefore, subendothelial accumulation of FN matrix is a cause, not an effect, of endothelial monolayer disorganization and leakiness. Regulating FN accumulation in the subendothelial space could be an important target for controlling progression of fibrosis and related diseases.
Collapse
Affiliation(s)
- Henry A. Resnikoff
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544
| | | |
Collapse
|
15
|
Fakhri N, Khalili A, Sachlos T, Rezai P. Fabrication of Porous Collagen Scaffolds Containing Embedded Channels with Collagen Membrane Linings. MICROMACHINES 2024; 15:1031. [PMID: 39203682 PMCID: PMC11356104 DOI: 10.3390/mi15081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024]
Abstract
Tissues and organs contain an extracellular matrix (ECM). In the case of blood vessels, endothelium cells are anchored to a specialized basement membrane (BM) embedded inside the interstitial matrix (IM). We introduce a multi-structural collagen-based scaffold with embedded microchannels that mimics in vivo structures within vessels. Our scaffold consists of two parts, each containing two collagen layers, i.e., a 3D porous collagen layer analogous to IM lined with a thin 2D collagen film resembling the BM. Enclosed microchannels were fabricated using contact microprinting. Microchannel test structures with different sizes ranging from 300 to 800 µm were examined for their fabrication reproducibility. The heights and perimeters of the fabricated microchannels were ~20% less than their corresponding values in the replication PDMS mold; however, microchannel widths were significantly closer to their replica dimensions. The stiffness, permeability, and pore size properties of the 2D and 3D collagen layers were measured. The permeability of the 2D collagen film was negligible, making it suitable for mimicking the BM of large blood vessels. A leakage test at various volumetric flow rates applied to the microchannels showed no discharge, thereby verifying the reliability of the proposed integrated 2D/3D collagen parts and the contact printing method used for bonding them in the scaffold. In the future, multi-cell culturing will be performed within the 3D porous collagen and against the 2D membrane inside the microchannel, hence preparing this scaffold for studying a variety of blood vessel-tissue interfaces. Also, thicker collagen scaffold tissues will be fabricated by stacking several layers of the proposed scaffold.
Collapse
Affiliation(s)
| | | | - Terry Sachlos
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
16
|
Li J, You D, Hu L, Yang Y, Gao S, Bai W. Identification and validation of basement membrane-associated gene AGRN as prognostic and immune-associated biomarkers in colorectal cancer patients. J Cell Mol Med 2024; 28:e70010. [PMID: 39183444 PMCID: PMC11345205 DOI: 10.1111/jcmm.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/16/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (COCA) has a poor prognosis, with growing evidence implicating basement membranes (BMs) in cancer progression. Our goal was to investigate the role and predictive significance of BMs in COCA patients. We obtained BMs-related genes from cutting-edge research and used TCGA and GTEx databases for mRNA expression and patient information. Cox regression and LASSO regression were used for prognostic gene selection and risk model construction. We compared prognosis using Kaplan-Meier analysis and examined drug sensitivity differences. The CMAP dataset identified potential small molecule drugs. In vitro tests involved suppressing a crucial gene to observe its impact on tumour metastasis. We developed a 12 BMs-based approach, finding it to be an independent prognostic factor. Functional analysis showed BMs concentrated in cancer-associated pathways, correlating with immune cell infiltration and immune checkpoint activation. High-risk individuals exhibited increased drug sensitivity. AGRN levels were linked to decreased progression-free survival (p < 0.001). AGRN knockdown suppressed tumour growth and metastasis. Our study offers new perspectives on BMs in COCA, concluding that AGRN is a dependable biomarker for patient survival and prognosis.
Collapse
Affiliation(s)
- Jianrong Li
- Department of General Surgery Sciences, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Daofeng You
- Emergency Department of First Hospital Affiliated to Hebei Medical UniversityShijiazhuangChina
| | - Linjie Hu
- Department of General Surgery Sciences, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Yusi Yang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences Tongji Shanxi HospitalTaiyuanChina
| | - Sheng Gao
- Department of General Surgery Sciences, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| | - Wenqi Bai
- Department of General Surgery Sciences, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer HospitalChinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
17
|
Yurchenco PD, Kulczyk AW. Polymerizing laminins in development, health, and disease. J Biol Chem 2024; 300:107429. [PMID: 38825010 PMCID: PMC11260871 DOI: 10.1016/j.jbc.2024.107429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/12/2024] [Accepted: 05/26/2024] [Indexed: 06/04/2024] Open
Abstract
Polymerizing laminins are multi-domain basement membrane (BM) glycoproteins that self-assemble into cell-anchored planar lattices to establish the initial BM scaffold. Nidogens, collagen-IV and proteoglycans then bind to the scaffold at different domain loci to create a mature BM. The LN domains of adjacent laminins bind to each other to form a polymer node, while the LG domains attach to cytoskeletal-anchoring integrins and dystroglycan, as well as to sulfatides and heparan sulfates. The polymer node, the repeating unit of the polymer scaffold, is organized into a near-symmetrical triskelion. The structure, recently solved by cryo-electron microscopy in combination with AlphaFold2 modeling and biochemical studies, reveals how the LN surface residues interact with each other and how mutations cause failures of self-assembly in an emerging group of diseases, the LN-lamininopathies, that include LAMA2-related dystrophy and Pierson syndrome.
Collapse
Affiliation(s)
- Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers University, Piscataway, New Jersey, USA.
| | - Arkadiusz W Kulczyk
- Department of Biochemistry and Microbiology, Institute for Quantitative Biomedicine, Rutgers University, Piscataway, New Jersey, USA
| |
Collapse
|
18
|
Choi SY, Kim HJ, Hwang S, Park J, Park J, Lee JW, Son KH. The Modulation of Respiratory Epithelial Cell Differentiation by the Thickness of an Electrospun Poly-ε-Carprolactone Mesh Mimicking the Basement Membrane. Int J Mol Sci 2024; 25:6650. [PMID: 38928356 PMCID: PMC11203971 DOI: 10.3390/ijms25126650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
The topology of the basement membrane (BM) affects cell physiology and pathology, and BM thickening is associated with various chronic lung diseases. In addition, the topology of commercially available poly (ethylene terephthalate) (PET) membranes, which are used in preclinical in vitro models, differs from that of the human BM, which has a fibrous and elastic structure. In this study, we verified the effect of BM thickness on the differentiation of normal human bronchial epithelial (NHBE) cells. To evaluate whether the thickness of poly-ε-carprolactone (PCL) mesh affects the differentiation of NHBE cells, cells were grown on thin- (6-layer) and thick-layer (80-layer) meshes consisting of electrospun PCL nanofibers using an air-liquid interface (ALI) cell culture system. It was found that the NHBE cells formed a normal pseudostratified epithelium composed of ciliated, goblet, and basal cells on the thin-layer PCL mesh; however, goblet cell hyperplasia was observed on the thick-layer PCL mesh. Differentiated NHBE cells cultured on the thick-layer PCL mesh also demonstrated increased epithelial-mesenchymal transition (EMT) compared to those cultured on the thin-layer PCL mesh. In addition, expression of Sox9, nuclear factor (NF)-κB, and oxidative stress-related markers, which are also associated with goblet cell hyperplasia, was increased in the differentiated NHBE cells cultured on the thick-layer PCL mesh. Thus, the use of thick electrospun PCL mesh led to NHBE cells differentiating into hyperplastic goblet cells via EMT and the oxidative stress-related signaling pathway. Therefore, the topology of the BM, for example, thickness, may affect the differentiation direction of human bronchial epithelial cells.
Collapse
Affiliation(s)
- Seon Young Choi
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Hyun Joo Kim
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Soyoung Hwang
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| | - Jangho Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
| | - Jungkyu Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
| | - Jin Woo Lee
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon 21999, Republic of Korea; (J.P.); (J.P.)
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
| | - Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gachon University Gil Medical Center, College of Medicine, Gachon University, Incheon 21565, Republic of Korea; (S.Y.C.); (H.J.K.); (S.H.)
| |
Collapse
|
19
|
Rosner M, Hengstschläger M. Oct4 controls basement membrane development during human embryogenesis. Dev Cell 2024; 59:1439-1456.e7. [PMID: 38579716 DOI: 10.1016/j.devcel.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/02/2024] [Accepted: 03/08/2024] [Indexed: 04/07/2024]
Abstract
Basement membranes (BMs) are sheet-like structures of extracellular matrix (ECM) that provide structural support for many tissues and play a central role in signaling. They are key regulators of cell behavior and tissue functions, and defects in their assembly or composition are involved in numerous human diseases. Due to the differences between human and animal embryogenesis, ethical concerns, legal constraints, the scarcity of human tissue material, and the inaccessibility of the in vivo condition, BM regulation during human embryo development has remained elusive. Using the post-implantation amniotic sac embryoid (PASE), we delineate BM assembly upon post-implantation development and BM disassembly during primitive streak (PS) cell dissemination. Further, we show that the transcription factor Oct4 regulates the expression of BM structural components and receptors and controls BM development by regulating Akt signaling and the small GTPase Rac1. These results represent a relevant step toward a more comprehensive understanding of early human development.
Collapse
Affiliation(s)
- Margit Rosner
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna 1090, Austria
| | - Markus Hengstschläger
- Institute of Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
20
|
Morales EA, Wang S. Salivary gland developmental mechanics. Curr Top Dev Biol 2024; 160:1-30. [PMID: 38937029 DOI: 10.1016/bs.ctdb.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
The salivary gland undergoes branching morphogenesis to elaborate into a tree-like structure with numerous saliva-secreting acinar units, all joined by a hierarchical ductal system. The expansive epithelial surface generated by branching morphogenesis serves as the structural basis for the efficient production and delivery of saliva. Here, we elucidate the process of salivary gland morphogenesis, emphasizing the role of mechanics. Structurally, the developing salivary gland is characterized by a stratified epithelium tightly encased by the basement membrane, which is in turn surrounded by a mesenchyme consisting of a dense network of interstitial matrix and mesenchymal cells. Diverse cell types and extracellular matrices bestow this developing organ with organized, yet spatially varied mechanical properties. For instance, the surface epithelial sheet of the bud is highly fluidic due to its high cell motility and weak cell-cell adhesion, rendering it highly pliable. In contrast, the inner core of the bud is more rigid, characterized by reduced cell motility and strong cell-cell adhesion, which likely provide structural support for the tissue. The interactions between the surface epithelial sheet and the inner core give rise to budding morphogenesis. Furthermore, the basement membrane and the mesenchyme offer mechanical constraints that could play a pivotal role in determining the higher-order architecture of a fully mature salivary gland.
Collapse
Affiliation(s)
- E Angelo Morales
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Shaohe Wang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States.
| |
Collapse
|
21
|
Martins B, Pires M, Ambrósio AF, Girão H, Fernandes R. Contribution of extracellular vesicles for the pathogenesis of retinal diseases: shedding light on blood-retinal barrier dysfunction. J Biomed Sci 2024; 31:48. [PMID: 38730462 PMCID: PMC11088087 DOI: 10.1186/s12929-024-01036-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
Retinal degenerative diseases, including diabetic retinopathy (DR) and age-related macular degeneration (AMD), loom as threats to vision, causing detrimental effects on the structure and function of the retina. Central to understanding these diseases, is the compromised state of the blood-retinal barrier (BRB), an effective barrier that regulates the influx of immune and inflammatory components. Whether BRB breakdown initiates retinal distress, or is a consequence of disease progression, remains enigmatic. Nevertheless, it is an indication of retinal dysfunction and potential vision loss.The intricate intercellular dialogues among retinal cell populations remain unintelligible in the complex retinal milieu, under conditions of inflammation and oxidative stress. The retina, a specialized neural tissue, sustains a ceaseless demand for oxygen and nutrients from two vascular networks. The BRB orchestrates the exchange of molecules and fluids within this specialized region, comprising the inner BRB (iBRB) and the outer BRB (oBRB). Extracellular vesicles (EVs) are small membranous structures, and act as messengers facilitating intercellular communication in this milieu.EVs, both from retinal and peripheral immune cells, increase complexity to BRB dysfunction in DR and AMD. Laden with bioactive cargoes, these EVs can modulate the retinal microenvironment, influencing disease progression. Our review delves into the multifaceted role of EVs in retinal degenerative diseases, elucidating the molecular crosstalk they orchestrate, and their microRNA (miRNA) content. By shedding light on these nanoscale messengers, from their biogenesis, release, to interaction and uptake by target cells, we aim to deepen the comprehension of BRB dysfunction and explore their therapeutic potential, therefore increasing our understanding of DR and AMD pathophysiology.
Collapse
Affiliation(s)
- Beatriz Martins
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - Maria Pires
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
| | - António Francisco Ambrósio
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal
| | - Henrique Girão
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal
| | - Rosa Fernandes
- University Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, 3000- 548, Portugal.
- University of Coimbra, Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, Coimbra, 3000-548, Portugal.
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, 3004-531, Portugal.
- Clinical Academic Center of Coimbra (CACC), Coimbra, 3004-561, Portugal.
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, 3000-548, Portugal.
| |
Collapse
|
22
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
23
|
Jiang S, Wise SG, Kovacic JC, Rnjak-Kovacina J, Lord MS. Biomaterials containing extracellular matrix molecules as biomimetic next-generation vascular grafts. Trends Biotechnol 2024; 42:369-381. [PMID: 37852854 DOI: 10.1016/j.tibtech.2023.09.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/18/2023] [Accepted: 09/19/2023] [Indexed: 10/20/2023]
Abstract
The performance of synthetic biomaterial vascular grafts for the bypass of stenotic and dysfunctional blood vessels remains an intractable challenge in small-diameter applications. The functionalization of biomaterials with extracellular matrix (ECM) molecules is a promising approach because these molecules can regulate multiple biological processes in vascular tissues. In this review, we critically examine emerging approaches to ECM-containing vascular graft biomaterials and explore opportunities for future research and development toward clinical use.
Collapse
Affiliation(s)
- Shouyuan Jiang
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Steven G Wise
- School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, NSW 2006, Australia; Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; The University of Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia
| | - Jason C Kovacic
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW 2010, Australia; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
24
|
Thomasy SM, Leonard BC, Greiner MA, Skeie JM, Raghunathan VK. Squishy matters - Corneal mechanobiology in health and disease. Prog Retin Eye Res 2024; 99:101234. [PMID: 38176611 PMCID: PMC11193890 DOI: 10.1016/j.preteyeres.2023.101234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
The cornea, as a dynamic and responsive tissue, constantly interacts with mechanical forces in order to maintain its structural integrity, barrier function, transparency and refractive power. Cells within the cornea sense and respond to various mechanical forces that fundamentally regulate their morphology and fate in development, homeostasis and pathophysiology. Corneal cells also dynamically regulate their extracellular matrix (ECM) with ensuing cell-ECM crosstalk as the matrix serves as a dynamic signaling reservoir providing biophysical and biochemical cues to corneal cells. Here we provide an overview of mechanotransduction signaling pathways then delve into the recent advances in corneal mechanobiology, focusing on the interplay between mechanical forces and responses of the corneal epithelial, stromal, and endothelial cells. We also identify species-specific differences in corneal biomechanics and mechanotransduction to facilitate identification of optimal animal models to study corneal wound healing, disease, and novel therapeutic interventions. Finally, we identify key knowledge gaps and therapeutic opportunities in corneal mechanobiology that are pressing for the research community to address especially pertinent within the domains of limbal stem cell deficiency, keratoconus and Fuchs' endothelial corneal dystrophy. By furthering our understanding corneal mechanobiology, we can contextualize discoveries regarding corneal diseases as well as innovative treatments for them.
Collapse
Affiliation(s)
- Sara M Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States; California National Primate Research Center, Davis, CA, United States.
| | - Brian C Leonard
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California - Davis, Davis, CA, United States; Department of Ophthalmology & Vision Science, School of Medicine, University of California - Davis, Davis, CA, United States
| | - Mark A Greiner
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | - Jessica M Skeie
- Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, United States; Iowa Lions Eye Bank, Coralville, IA, United States
| | | |
Collapse
|
25
|
Radzki D, Negri A, Kusiak A, Obuchowski M. Matrix Metalloproteinases in the Periodontium-Vital in Tissue Turnover and Unfortunate in Periodontitis. Int J Mol Sci 2024; 25:2763. [PMID: 38474009 DOI: 10.3390/ijms25052763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The extracellular matrix (ECM) is a complex non-cellular three-dimensional macromolecular network present within all tissues and organs, forming the foundation on which cells sit, and composed of proteins (such as collagen), glycosaminoglycans, proteoglycans, minerals, and water. The ECM provides a fundamental framework for the cellular constituents of tissue and biochemical support to surrounding cells. The ECM is a highly dynamic structure that is constantly being remodeled. Matrix metalloproteinases (MMPs) are among the most important proteolytic enzymes of the ECM and are capable of degrading all ECM molecules. MMPs play a relevant role in physiological as well as pathological processes; MMPs participate in embryogenesis, morphogenesis, wound healing, and tissue remodeling, and therefore, their impaired activity may result in several problems. MMP activity is also associated with chronic inflammation, tissue breakdown, fibrosis, and cancer invasion and metastasis. The periodontium is a unique anatomical site, composed of a variety of connective tissues, created by the ECM. During periodontitis, a chronic inflammation affecting the periodontium, increased presence and activity of MMPs is observed, resulting in irreversible losses of periodontal tissues. MMP expression and activity may be controlled in various ways, one of which is the inhibition of their activity by an endogenous group of tissue inhibitors of metalloproteinases (TIMPs), as well as reversion-inducing cysteine-rich protein with Kazal motifs (RECK).
Collapse
Affiliation(s)
- Dominik Radzki
- Department of Periodontology and Oral Mucosa Diseases, Faculty of Medicine, Medical University of Gdańsk, 80-208 Gdańsk, Poland
- Division of Molecular Bacteriology, Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Alessandro Negri
- Division of Molecular Bacteriology, Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Aida Kusiak
- Department of Periodontology and Oral Mucosa Diseases, Faculty of Medicine, Medical University of Gdańsk, 80-208 Gdańsk, Poland
| | - Michał Obuchowski
- Division of Molecular Bacteriology, Institute of Medical Biotechnology and Experimental Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| |
Collapse
|
26
|
Antonyshyn JA, MacQuarrie KD, McFadden MJ, Gramolini AO, Hofer SOP, Santerre JP. Paracrine cross-talk between human adipose tissue-derived endothelial cells and perivascular cells accelerates the endothelialization of an electrospun ionomeric polyurethane scaffold. Acta Biomater 2024; 175:214-225. [PMID: 38158104 DOI: 10.1016/j.actbio.2023.12.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The ex vivo endothelialization of small diameter vascular prostheses can prolong their patency. Here, we demonstrate that heterotypic interactions between human adipose tissue-derived endothelial cells and perivascular cells can be exploited to accelerate the endothelialization of an electrospun ionomeric polyurethane scaffold. The scaffold was used to physically separate endothelial cells from perivascular cells to prevent their diffuse neo-intimal hyperplasia and spontaneous tubulogenesis, yet enable their paracrine cross-talk to accelerate the integration of the endothelial cells into a temporally stable endothelial lining of a continuous, elongated, and aligned morphology. Perivascular cells stimulated endothelial basement membrane protein production and suppressed their angiogenic and inflammatory activation to accelerate this biomimetic morphogenesis of the endothelium. These findings demonstrate the feasibility and underscore the value of exploiting heterotypic interactions between endothelial cells and perivascular cells for the fabrication of an endothelial lining intended for small diameter arterial reconstruction. STATEMENT OF SIGNIFICANCE: Adipose tissue is an abundant, accessible, and uniquely dispensable source of endothelial cells and perivascular cells for vascular tissue engineering. While their spontaneous self-assembly into microvascular networks is routinely exploited for the vascularization of engineered tissues, it threatens the temporal stability of an endothelial lining intended for small diameter arterial reconstruction. Here, we demonstrate that an electrospun polyurethane scaffold can be used to physically separate endothelial cells from perivascular cells to prevent their spontaneous capillary morphogenesis, yet enable their cross-talk to promote the formation of a stable endothelium. Our findings demonstrate the feasibility of engineering an endothelial lining from human adipose tissue, poising it for the rapid ex vivo endothelialization of small diameter vascular prostheses in an autologous, patient-specific manner.
Collapse
Affiliation(s)
- Jeremy A Antonyshyn
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada
| | - Kate D MacQuarrie
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada
| | - Meghan J McFadden
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Stefan O P Hofer
- Division of Plastic, Reconstructive, and Aesthetic Surgery, University of Toronto, Toronto, Canada; Departments of Surgery and Surgical Oncology, University Health Network, Toronto, Canada
| | - J Paul Santerre
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, Canada; Faculty of Dentistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
27
|
Grewal S, Gonçalves de Andrade E, Kofoed RH, Matthews PM, Aubert I, Tremblay MÈ, Morse SV. Using focused ultrasound to modulate microglial structure and function. Front Cell Neurosci 2023; 17:1290628. [PMID: 38164436 PMCID: PMC10757935 DOI: 10.3389/fncel.2023.1290628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Transcranial focused ultrasound (FUS) has the unique ability to target regions of the brain with high spatial precision, in a minimally invasive manner. Neuromodulation studies have shown that FUS can excite or inhibit neuronal activity, demonstrating its tremendous potential to improve the outcome of neurological diseases. Recent evidence has also shed light on the emerging promise that FUS has, with and without the use of intravenously injected microbubbles, in modulating the blood-brain barrier and the immune cells of the brain. As the resident immune cells of the central nervous system, microglia are at the forefront of the brain's maintenance and immune defense. Notably, microglia are highly dynamic and continuously survey the brain parenchyma by extending and retracting their processes. This surveillance activity aids microglia in performing key physiological functions required for brain activity and plasticity. In response to stressors, microglia rapidly alter their cellular and molecular profile to help facilitate a return to homeostasis. While the underlying mechanisms by which both FUS and FUS + microbubbles modify microglial structure and function remain largely unknown, several studies in adult mice have reported changes in the expression of the microglia/macrophage marker ionized calcium binding adaptor molecule 1, and in their phagocytosis, notably of protein aggregates, such as amyloid beta. In this review, we discuss the demonstrated and putative biological effects of FUS and FUS + microbubbles in modulating microglial activities, with an emphasis on the key cellular and molecular changes observed in vitro and in vivo across models of brain health and disease. Understanding how this innovative technology can modulate microglia paves the way for future therapeutic strategies aimed to promote beneficial physiological microglial roles, and prevent or treat maladaptive responses.
Collapse
Affiliation(s)
- Sarina Grewal
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Center for Experimental Neuroscience-CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Paul M. Matthews
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sophie V. Morse
- Department of Bioengineering, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
Cohen R, Baruch ES, Cabilly I, Shapira A, Dvir T. Modified ECM-Based Bioink for 3D Printing of Multi-Scale Vascular Networks. Gels 2023; 9:792. [PMID: 37888365 PMCID: PMC10606913 DOI: 10.3390/gels9100792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/18/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023] Open
Abstract
The survival and function of tissues depend on appropriate vascularization. Blood vessels of the tissues supply oxygen, and nutrients and remove waste and byproducts. Incorporating blood vessels into engineered tissues is essential for overcoming diffusion limitations, improving tissue function, and thus facilitating the fabrication of thick tissues. Here, we present a modified ECM bioink, with enhanced mechanical properties and endothelial cell-specific adhesion motifs, to serve as a building material for 3D printing of a multiscale blood vessel network. The bioink is composed of natural ECM and alginate conjugated with a laminin adhesion molecule motif (YIGSR). The hybrid hydrogel was characterized for its mechanical properties, biochemical content, and ability to interact with endothelial cells. The pristine and modified hydrogels were mixed with induced pluripotent stem cells derived endothelial cells (iPSCs-ECs) and used to print large blood vessels with capillary beds in between.
Collapse
Affiliation(s)
- Roni Cohen
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ester-Sapir Baruch
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Department of Materials Science and Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Itai Cabilly
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
| | - Assaf Shapira
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
| | - Tal Dvir
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Shmunis School of Biomedicine and Cancer Research, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel; (E.-S.B.); (I.C.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol Center for Regenerative Biotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
29
|
Wang D, Brady T, Santhanam L, Gerecht S. The extracellular matrix mechanics in the vasculature. NATURE CARDIOVASCULAR RESEARCH 2023; 2:718-732. [PMID: 39195965 DOI: 10.1038/s44161-023-00311-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/20/2023] [Indexed: 08/29/2024]
Abstract
Mechanical stimuli from the extracellular matrix (ECM) modulate vascular differentiation, morphogenesis and dysfunction of the vasculature. With innovation in measurements, we can better characterize vascular microenvironment mechanics in health and disease. Recent advances in material sciences and stem cell biology enable us to accurately recapitulate the complex and dynamic ECM mechanical microenvironment for in vitro studies. These biomimetic approaches help us understand the signaling pathways in disease pathologies, identify therapeutic targets, build tissue replacement and activate tissue regeneration. This Review analyzes how ECM mechanics regulate vascular homeostasis and dysfunction. We highlight approaches to examine ECM mechanics at tissue and cellular levels, focusing on how mechanical interactions between cells and the ECM regulate vascular phenotype, especially under certain pathological conditions. Finally, we explore the development of biomaterials to emulate, measure and alter the physical microenvironment of pathological ECM to understand cell-ECM mechanical interactions toward the development of therapeutics.
Collapse
Affiliation(s)
- Dafu Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Travis Brady
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
30
|
Abstract
Angiogenic sprouting, the formation of new blood vessels from pre-existing vasculature, is tightly regulated by the properties of the surrounding tissue microenvironment. Although the extracellular matrix has been shown to be a major regulator of this process, it is not clear how individual biochemical and mechanical properties influence endothelial cell sprouting. This information gap is largely due to the lack of suitable in vitro models that recapitulate angiogenic sprouting in a 3D environment with independent control over matrix properties. Here, we present protocols for the preparation of endothelial cell spheroid-laden synthetic, dextran-based hydrogels, which serve as a highly tunable 3D scaffold. The adjustment of the hydrogels' adhesiveness, stiffness, and degradability is demonstrated in detail. Finally, we describe assays to elucidate how individual matrix properties regulate angiogenic sprouting, including their analysis by immunofluorescence staining and imaging. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Synthesis of methacrylated dextran (DexMA) Basic Protocol 2: Generation of endothelial cell spheroids in microwells Basic Protocol 3: Endothelial cell sprouting in hydrogels of tunable stiffness Basic Protocol 4: Endothelial cell sprouting in hydrogels of tunable adhesiveness Basic Protocol 5: Endothelial cell sprouting in hydrogels of tunable degradability Basic Protocol 6: Imaging of endothelial cell spheroid-laden hydrogels Support Protocol 1: Preparation of pro-angiogenic cocktail for endothelial cell sprouting.
Collapse
Affiliation(s)
- Giuseppe Trapani
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Martin Sebastian Weiß
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
- Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
31
|
Jain P, Rimal R, Möller M, Singh S. Topographical influence of electrospun basement membrane mimics on formation of cellular monolayer. Sci Rep 2023; 13:8382. [PMID: 37225757 DOI: 10.1038/s41598-023-34934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 05/10/2023] [Indexed: 05/26/2023] Open
Abstract
Functional unit of many organs like lung, kidney, intestine, and eye have their endothelial and epithelial monolayers physically separated by a specialized extracellular matrix called the basement membrane. The intricate and complex topography of this matrix influences cell function, behavior and overall homeostasis. In vitro barrier function replication of such organs requires mimicking of these native features on an artificial scaffold system. Apart from chemical and mechanical features, the choice of nano-scale topography of the artificial scaffold is integral, however its influence on monolayer barrier formation is unclear. Though studies have reported improved single cell adhesion and proliferation in presence of pores or pitted topology, corresponding influence on confluent monolayer formation is not well reported. In this work, basement membrane mimic with secondary topographical cues is developed and its influence on single cells and their monolayers is investigated. We show that single cells cultured on fibers with secondary cues form stronger focal adhesions and undergo increased proliferation. Counterintuitively, absence of secondary cues promoted stronger cell-cell interaction in endothelial monolayers and promoted formation of integral tight barriers in alveolar epithelial monolayers. Overall, this work highlights the importance of choice of scaffold topology to develop basement barrier function in in vitro models.
Collapse
Affiliation(s)
- Puja Jain
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Rahul Rimal
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany
| | - Martin Möller
- DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074, Aachen, Germany
| | - Smriti Singh
- Max Planck Institute for Medical Research (MPImF), Jahnstrasse 29, 69120, Heidelberg, Germany.
| |
Collapse
|
32
|
Jandl K, Radic N, Zeder K, Kovacs G, Kwapiszewska G. Pulmonary vascular fibrosis in pulmonary hypertension - The role of the extracellular matrix as a therapeutic target. Pharmacol Ther 2023; 247:108438. [PMID: 37210005 DOI: 10.1016/j.pharmthera.2023.108438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/03/2023] [Accepted: 05/15/2023] [Indexed: 05/22/2023]
Abstract
Pulmonary hypertension (PH) is a condition characterized by changes in the extracellular matrix (ECM) deposition and vascular remodeling of distal pulmonary arteries. These changes result in increased vessel wall thickness and lumen occlusion, leading to a loss of elasticity and vessel stiffening. Clinically, the mechanobiology of the pulmonary vasculature is becoming increasingly recognized for its prognostic and diagnostic value in PH. Specifically, the increased vascular fibrosis and stiffening resulting from ECM accumulation and crosslinking may be a promising target for the development of anti- or reverse-remodeling therapies. Indeed, there is a huge potential in therapeutic interference with mechano-associated pathways in vascular fibrosis and stiffening. The most direct approach is aiming to restore extracellular matrix homeostasis, by interference with its production, deposition, modification and turnover. Besides structural cells, immune cells contribute to the level of ECM maturation and degradation by direct cell-cell contact or the release of mediators and proteases, thereby opening a huge avenue to target vascular fibrosis via immunomodulation approaches. Indirectly, intracellular pathways associated with altered mechanobiology, ECM production, and fibrosis, offer a third option for therapeutic intervention. In PH, a vicious cycle of persistent activation of mechanosensing pathways such as YAP/TAZ initiates and perpetuates vascular stiffening, and is linked to key pathways disturbed in PH, such as TGF-beta/BMPR2/STAT. Together, this complexity of the regulation of vascular fibrosis and stiffening in PH allows the exploration of numerous potential therapeutic interventions. This review discusses connections and turning points of several of these interventions in detail.
Collapse
Affiliation(s)
- Katharina Jandl
- Division of Pharmacology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria.
| | - Nemanja Radic
- Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria
| | - Katarina Zeder
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Gabor Kovacs
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Graz, Austria; Division of Physiology, Otto Loewi Research Center, Medical University Graz, Graz, Austria; Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany
| |
Collapse
|
33
|
Bando Y, Nagasaka A, Onozawa G, Sakiyama K, Owada Y, Amano O. Integrin expression and extracellular matrix adhesion of septoclasts, pericytes, and endothelial cells at the chondro-osseous junction and the metaphysis of the proximal tibia in young mice. J Anat 2023; 242:831-845. [PMID: 36602038 PMCID: PMC10093157 DOI: 10.1111/joa.13820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/16/2022] [Accepted: 12/19/2022] [Indexed: 01/06/2023] Open
Abstract
We previously reported that septoclasts, which are uncalcified growth plate (GP) cartilage matrix-resorbing cells, are derived from pericytes surrounding capillary endothelial cells. Resorption of the GP is assumed to be regulated synchronously by septoclasts, pericytes, and endothelial cells. To reveal the contribution of the extracellular matrix (ECM) to the regulatory mechanisms of septoclastic cartilage resorption, we investigated the spatial correlation between the cells and the ECM in the GP matrix and basement membrane (BM) and investigated the expression of integrins-ECM receptors-in the cells. Septoclasts attached to the transverse septa containing collagen-II/-X at the tip of their processes and to the longitudinal septa containing collagen-II/-X at the spine-like processes extending from their bodies and processes. Collagen-IV and laminin α4 in the BM were sparsely detected between septoclasts and capillary endothelial cells at the chondro-osseous junction (COJ) and were absent in the outer surface of pericytes at the metaphysis. Integrin α1/α2, integrin α1, and integrin α2/α6 were detected in the cell membranes of septoclasts, pericytes, and endothelial cells, respectively. These results suggest that the adhesion between septoclasts and the cartilage ECM forming the scaffolds for cartilage resorption and migration is provided by integrin α2-collagen-II/-X interaction and that the adhesions between the BM and pericytes or endothelial cells are mediated by integrin α1-collagen-IV and integrin α2/α6-laminin interaction, respectively.
Collapse
Affiliation(s)
- Yasuhiko Bando
- Division of HistologyMeikai University School of DentistrySaitamaJapan
| | - Arata Nagasaka
- Division of HistologyMeikai University School of DentistrySaitamaJapan
| | - Go Onozawa
- Division of HistologyMeikai University School of DentistrySaitamaJapan
- Division of Oral and Maxillofacial SurgeryMeikai University School of DentistrySaitamaJapan
| | - Koji Sakiyama
- Division of AnatomyMeikai University School of DentistrySaitamaJapan
| | - Yuji Owada
- Department of Organ AnatomyTohoku University Graduate School of MedicineSendaiJapan
| | - Osamu Amano
- Division of HistologyMeikai University School of DentistrySaitamaJapan
| |
Collapse
|
34
|
Knopp RC, Erickson MA, Rhea EM, Reed MJ, Banks WA. Cellular senescence and the blood-brain barrier: Implications for aging and age-related diseases. Exp Biol Med (Maywood) 2023; 248:399-411. [PMID: 37012666 PMCID: PMC10281623 DOI: 10.1177/15353702231157917] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
Abstract
The blood-brain barrier (BBB) is a critical physiochemical interface that regulates communication between the brain and blood. It is comprised of brain endothelial cells which regulate the BBB's barrier and interface properties and is surrounded by supportive brain cell types including pericytes and astrocytes. Recent reports have suggested that the BBB undergoes dysfunction during normative aging and in disease. In this review, we consider the effect of cellular senescence, one of the nine hallmarks of aging, on the BBB. We first characterize known normative age-related changes at the BBB, and then evaluate changes in neurodegenerative diseases, with an emphasis on if/how cellular senescence is influencing these changes. We then discuss what insight has been gained from in vitro and in vivo studies of cellular senescence at the BBB. Finally, we evaluate mechanisms by which cellular senescence in peripheral pathologies can indirectly or directly affect BBB function.
Collapse
Affiliation(s)
- Rachel C Knopp
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Michelle A Erickson
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - Elizabeth M Rhea
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - May J Reed
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| | - William A Banks
- Veterans Affairs Puget Sound Health Care
System, Geriatrics Research Education and Clinical Center (GRECC), Seattle, WA 98108,
USA
- Department of Medicine, Division of
Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA
98195, USA
| |
Collapse
|
35
|
Costa D, Andreucci M, Ielapi N, Serraino GF, Mastroroberto P, Bracale UM, Serra R. Vascular Biology of arterial aneurysms. Ann Vasc Surg 2023:S0890-5096(23)00225-X. [PMID: 37068624 DOI: 10.1016/j.avsg.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/19/2023]
Abstract
OBJECTIVE This review aims to analyze biomolecular and cellular events responsible for arterial aneurysm formation with particular attention to vascular remodeling that determines the initiation and the progression of arterial aneurysm, till rupture. METHODS This review was conducted searching libraries such as Web of Science, Scopus, ScienceDirect and Medline. Used keywords with various combinations were: "arterial aneurysms", "biology", "genetics", "proteomics", "molecular", "pathophysiology" and extracellular matrix" RESULTS: There are several genetic alterations responsible of syndromic and non-syndromic disease that predispose to aneurysm formation. ECM imbalance, mainly due to the alteration of vascular smooth muscle cells (VSMCs) homeostasis, overexpression of metalloproteinases (MPs) and cytokines activation, determines weakness of the arterial wall that dilates thus causing aneurysmal disease. Altered mechanotransduction in the ECM may also trigger and sustain anomalous cellular and biochemical signaling. Different cell population such as VSMCs, macrophages, perivascular adipose tissue (PVAT) cells, vascular wall resident stem cells (VWRSCs) are all involved at different levels CONCLUSIONS: Improving knowledge in vascular biology may help researchers and physicians in better targeting aneurysmal disease in order to better prevent and better treat such important disease.
Collapse
Affiliation(s)
- Davide Costa
- Department of Law, Economics and Sociology, University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy; Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology. University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy
| | - Michele Andreucci
- Department of Health Sciences. University "Magna Graecia" of Catanzaro. 88100, Catanzaro, Italy
| | - Nicola Ielapi
- Department of Public Health and Infectious Disease, "Sapienza" University of Rome, 00185, Rome, Italy
| | - Giuseppe Filiberto Serraino
- Department of Experimental and Clinical Medicine. University "Magna Graecia" of Catanzaro. 88100, Catanzaro, Italy
| | - Pasquale Mastroroberto
- Department of Experimental and Clinical Medicine. University "Magna Graecia" of Catanzaro. 88100, Catanzaro, Italy
| | | | - Raffaele Serra
- Interuniversity Center of Phlebolymphology (CIFL), International Research and Educational Program in Clinical and Experimental Biotechnology. University "Magna Graecia" of Catanzaro, 88100 Catanzaro, Italy; Department of Medical and Surgical Sciences. University "Magna Graecia" of Catanzaro. 88100, Catanzaro, Italy.
| |
Collapse
|
36
|
Zhang Z, Zhu H, Wang X, Lin S, Ruan C, Wang Q. A novel basement membrane-related gene signature for prognosis of lung adenocarcinomas. Comput Biol Med 2023; 154:106597. [PMID: 36708655 DOI: 10.1016/j.compbiomed.2023.106597] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/01/2022] [Accepted: 01/22/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) remains a global health concern with its poor prognosis and high mortality. Whether tumor cells invade through the basement membrane (BM) is the key factor to determine the prognosis of LUAD. This study aimed to identify the BM-related gene signatures to improve the overall prognosis of LUAD. MATERIALS & METHODS A series of bioinformatics analyses were conducted based on TCGA and GEO datasets. Unsupervised consistent cluster analysis was performed, and 500 LUAD patients were assigned to two different groups according to expressions of 222 BM-related genes. The differentially expressed genes (DEGs) between the two clusters were identified, and Lasso regression, ROC curve, univariate and multivariate Cox regression analyses and enrichment analysis were conducted. Besides, ssGSEA, CIBERSORT and ESTIMATE algorithmwere were employed to understand the relationship between the tumor microenvironment (TME) and risk scores. Moreover, single cell clustering and trajectory analyses were performed to further understand the significance of BM-related genes. Finally, qRT-PCR was used to verify the prognosis model. RESULTS A total of 31 prognostic BM-related genes were determined for LUAD, and a novel 17-mRNA prognostic model named BMsocre was successfully established to predict the overall survival of LUAD patients. The high BMscore group indicated worse prognosis. Seventeen DEGs were enriched mainly in metabolism, ECM-receptor interaction and immune response. In addition, the high-risk group showed higher TMB and lower immune score. The low-risk group had a better immunotherapeutic response where immune escape was less likely. The BMscore model was verified in our patient cohort. Furthermore, NELL2 was mainly expressed in clusters of T cells, and was identified to play a critical role in T-cell differentiation. CONCLUSIONS A novel BMscore model was successfully established and might be effective for providing guidance to LUAD therapy.
Collapse
Affiliation(s)
- Zhenxing Zhang
- Department of Thoracic and Maxillofacial Surgery (B7X), Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province, China
| | - Haoran Zhu
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi Province, China
| | - Xiaojun Wang
- Department of Thoracic Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province, China
| | - Shanan Lin
- Department of Thoracic Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province, China
| | - Chenjin Ruan
- Department of Thoracic Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province, China
| | - Qiang Wang
- Department of Thoracic Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang Province, China.
| |
Collapse
|
37
|
Hamrangsekachaee M, Wen K, Bencherif SA, Ebong EE. Atherosclerosis and endothelial mechanotransduction: current knowledge and models for future research. Am J Physiol Cell Physiol 2023; 324:C488-C504. [PMID: 36440856 PMCID: PMC10069965 DOI: 10.1152/ajpcell.00449.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/16/2022] [Accepted: 11/20/2022] [Indexed: 11/29/2022]
Abstract
Endothelium health is essential to the regulation of physiological vascular functions. Because of the critical capability of endothelial cells (ECs) to sense and transduce chemical and mechanical signals in the local vascular environment, their dysfunction is associated with a vast variety of vascular diseases and injuries, especially atherosclerosis and subsequent cardiovascular diseases. This review describes the mechanotransduction events that are mediated through ECs, the EC subcellular components involved, and the pathways reported to be potentially involved. Up-to-date research efforts involving in vivo animal models and in vitro biomimetic models are also discussed, including their advantages and drawbacks, with recommendations on future modeling approaches to aid the development of novel therapies targeting atherosclerosis and related cardiovascular diseases.
Collapse
Affiliation(s)
| | - Ke Wen
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
| | - Sidi A Bencherif
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
- Bioengineering Department, Northeastern University, Boston, Massachusetts
- Laboratoire de BioMécanique et BioIngénierie, UMR CNRS 7388, Sorbonne Universités, Université de Technologie of Compiègne, Compiègne, France
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Eno E Ebong
- Chemical Engineering Department, Northeastern University, Boston, Massachusetts
- Bioengineering Department, Northeastern University, Boston, Massachusetts
- Neuroscience Department, Albert Einstein College of Medicine, New York, New York
| |
Collapse
|
38
|
Ji W, Wu Z, Wen J, Tang H, Chen Z, Xue B, Tian Z, Ba Y, Zhang N, Wen X, Hou B. A simple method to isolate structurally and chemically intact brain vascular basement membrane for neural regeneration following traumatic brain injury. Biomater Res 2023; 27:2. [PMID: 36635718 PMCID: PMC9837976 DOI: 10.1186/s40824-023-00341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/02/2023] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND The brain vascular basement membrane (brain-VBM) is an important component of the brain extracellular matrix, and the three-dimensional structure of the cerebrovascular network nested with many cell-adhesive proteins may provide guidance for brain tissue regeneration. However, the potential of ability of brain-VBM to promote neural tissue regeneration has not been examined due to the technical difficulty of isolating intact brain-VBM. METHODS The present study developed a simple, effective method to isolate structurally and compositionally intact brain-VBM. Structural and component properties of the brain-VBM were characterized to confirm the technique. Seed cells were cocultured with brain-VBM in vitro to analyze biocompatibility and neurite extension. An experimental rat model of focal traumatic brain injury (TBI) induced by controlled cortical impact were conducted to further test the tissue regeneration ability of brain-VBM. RESULTS Brain-VBM isolated using genipin showed significantly improved mechanical properties, was easy to handle, supported high cell viability, exhibited strong cell adhesive properties, and promoted neurite extension and outgrowth. Further testing of the isolated brain-VBM transplanted at lesion sites in an experimental rat model of focal TBI demonstrated considerable promise for reconstructing a complete blood vessel network that filled in the lesion cavity and promoting repopulation of neural progenitor cells and neurons. CONCLUSION The technique allows isolation of intact brain-VBM as a 3D microvascular scaffold to support brain tissue regeneration following TBI and shows considerable promise for the production of naturally-derived biomaterials for neural tissue engineering.
Collapse
Affiliation(s)
- Wanqing Ji
- grid.410737.60000 0000 8653 1072Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623 China
| | - Zhiru Wu
- grid.412679.f0000 0004 1771 3402Department of Nephrology, Dongcheng branch of the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiaming Wen
- grid.410737.60000 0000 8653 1072Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623 China
| | - Hengxin Tang
- grid.79703.3a0000 0004 1764 3838Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| | - Zhuopeng Chen
- grid.12981.330000 0001 2360 039XDepartment of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 Guangdong Province China
| | - Bo Xue
- grid.268154.c0000 0001 2156 6140Shared Research Facilities, West Virginia University, 1306 Evansdale Drive, Morgantown, WV 26506 USA
| | - Zhenming Tian
- grid.12981.330000 0001 2360 039XDepartment of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 Guangdong Province China
| | - Yueyang Ba
- grid.12981.330000 0001 2360 039XDepartment of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 Guangdong Province China
| | - Ning Zhang
- grid.224260.00000 0004 0458 8737Department of Biomedical Engineering, Institute For Engineering and Medicine, Virginia Commonwealth University, Room 399, 601 West Main Street, Richmond, VA 23220 USA
| | - Xuejun Wen
- grid.224260.00000 0004 0458 8737Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 West Main Street, Richmond, VA 23220 USA
| | - Bo Hou
- grid.12981.330000 0001 2360 039XDepartment of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630 Guangdong Province China
| |
Collapse
|
39
|
Nanotopography and Microconfinement Impact on Primary Hippocampal Astrocyte Morphology, Cytoskeleton and Spontaneous Calcium Wave Signalling. Cells 2023; 12:cells12020293. [PMID: 36672231 PMCID: PMC9856934 DOI: 10.3390/cells12020293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Astrocytes' organisation affects the functioning and the fine morphology of the brain, both in physiological and pathological contexts. Although many aspects of their role have been characterised, their complex functions remain, to a certain extent, unclear with respect to their contribution to brain cell communication. Here, we studied the effects of nanotopography and microconfinement on primary hippocampal rat astrocytes. For this purpose, we fabricated nanostructured zirconia surfaces as homogenous substrates and as micrometric patterns, the latter produced by a combination of an additive nanofabrication and micropatterning technique. These engineered substrates reproduce both nanotopographical features and microscale geometries that astrocytes encounter in their natural environment, such as basement membrane topography, as well as blood vessels and axonal fibre topology. The impact of restrictive adhesion manifests in the modulation of several cellular properties of single cells (morphological and actin cytoskeletal changes) and the network organisation and functioning. Calcium wave signalling was observed only in astrocytes grown in confined geometries, with an activity enhancement in cells forming elongated agglomerates with dimensions typical of blood vessels or axon fibres. Our results suggest that calcium oscillation and wave propagation are closely related to astrocytic morphology and actin cytoskeleton organisation.
Collapse
|
40
|
Yu P, Zhang G, Hou B, Song E, Wen J, Ba Y, Zhu D, Wang G, Qin F. Effects of ECM proteins (laminin, fibronectin, and type IV collagen) on the biological behavior of Schwann cells and their roles in the process of remyelination after peripheral nerve injury. Front Bioeng Biotechnol 2023; 11:1133718. [PMID: 37034260 PMCID: PMC10080002 DOI: 10.3389/fbioe.2023.1133718] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Introduction: It is important to note that complete myelination and formation of myelinated fibers are essential for functional nerve regeneration after peripheral nerve injury (PNI). However, suboptimal myelin regeneration is common and can hinder ideal nerve regeneration. Therefore, it is important to closely monitor and support myelin regeneration in patients with PNI to achieve optimal outcomes. Methods: This study analyzed the effects of three extracellular matrix (ECM) proteins on Schwann cells (SCs) in the nerve regeneration environment, including their adhesion, proliferation, and migration. The study also explored the use of composite sodium alginate hydrogel neural scaffolds with ECM components and investigated the effects of ECM proteins on remyelination following peripheral nerve injury. Results: The results showed that laminin (LN), fibronectin (FN), and collagen Ⅳ (type IV Col) promoted the early adhesion of SCs in 2-dimensional culture but the ratios of early cell adhesion were quite different and the maintenance of cells' morphology by different ECM proteins were significantly different. In transwell experiment, the ability of LN and FN to induce the migration of SCs was obviously higher than that of type IV Col. An vitro co-culture model of SCs and dorsal root ganglia (DRG) neurons showed that LN promoted the transition of SCs to a myelinated state and the maturation of the myelin sheath, and increased the thickness of neurofilaments. Animal experiments showed that LN had superior effects in promoting myelin sheath formation, axon repair, and reaching an ideal G-ratio after injury compared to FN and Col IV. The situation of gastrocnemius atrophy was significantly better in the LN group. Notably, the thickness of the regenerated myelin sheaths in the type IV Col group was the thickest. Conclusion: In this experiment, we analyzed and compared the effects of LN, FN, and type IV Col on the biological behavior of SCs and their effects on remyelination after PNI and further clarified their unique roles in the process of remyelination. Further research is necessary to explore the underlying mechanisms.
Collapse
Affiliation(s)
- Peng Yu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guanhua Zhang
- Department of Cerebrovascular Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Bo Hou
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Enpeng Song
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jiaming Wen
- Department of Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yueyang Ba
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Donglin Zhu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- *Correspondence: Donglin Zhu, ; Gangwei Wang, ; Feng Qin,
| | - Gangwei Wang
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- *Correspondence: Donglin Zhu, ; Gangwei Wang, ; Feng Qin,
| | - Feng Qin
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- *Correspondence: Donglin Zhu, ; Gangwei Wang, ; Feng Qin,
| |
Collapse
|
41
|
Chen Y, Mao C, Gu R, Zhao R, Li W, Ma Z, Jia Y, Yu F, Luo J, Fu Y, Sun J, Kong W. Nidogen-2 is a Novel Endogenous Ligand of LGR4 to Inhibit Vascular Calcification. Circ Res 2022; 131:1037-1054. [PMID: 36354004 DOI: 10.1161/circresaha.122.321614] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Vascular calcification is closely related to the all-cause mortality of cardiovascular events. Basement membrane protein nidogen-2 is a key component of the vascular extracellular matrix microenvironment and we recently found it is pivotal for the maintenance of contractile phenotype in vascular smooth muscle cells (VSMCs). However, whether nidogen-2 is involved in VSMCs osteochondrogenic transition and vascular calcification remains unclear. METHODS VSMCs was treated with high-phosphate to study VSMC calcification in vitro. Three different mice models (5/6 nephrectomy-induced chronic renal failure, cholecalciferol-overload, and periadventitially administered with CaCl2) were used to study vascular calcification in vivo. Membrane protein interactome, coimmunoprecipitation, flow cytometric binding assay, surface plasmon resonance, G protein signaling, VSMCs calcium assays were performed to clarify the phenotype and elucidate the molecular mechanisms. RESULTS Nidogen-2 protein levels were significantly reduced in calcified VSMCs and aortas from mice in different vascular calcification model. Nidogen-2 deficiency exacerbated high-phosphate-induced VSMC calcification, whereas the addition of purified nidogen-2 protein markedly alleviated VSMC calcification in vitro. Nidogen-2-/- mice exhibited aggravated aorta calcification compared to wild-type (WT) mice in response to 5/6 nephrectomy, cholecalciferol-overload, and CaCl2 administration. Further unbiased coimmunoprecipitation and interactome analysis of purified nidogen-2 and membrane protein in VSMCs revealed that nidogen-2 directly binds to LGR4 (leucine-rich repeat G-protein-coupled receptor 4) with KD value 26.77 nM. LGR4 deficiency in VSMCs in vitro or in vivo abolished the protective effect of nidogen-2 on vascular calcification. Of interest, nidogen-2 biased activated LGR4-Gαq-PKCα (protein kinase Cα)-AMPKα1 (AMP-activated protein kinase α1) signaling to counteract VSMCs osteogenic transition and mineralization. CONCLUSIONS Nidogen-2 is a novel endogenous ligand of LGR4 that biased activated Gαq- PKCα-AMPKα1 signaling and inhibited vascular calcification.
Collapse
Affiliation(s)
- Yufei Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| | - Chenfeng Mao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Beijing Institute of Biotechnology, China (C.M.)
| | - Rui Gu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Beijing Institute of Biotechnology, China (C.M.)
| | - Rujia Zhao
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, China (R.Z., J.S.)
| | - Weihao Li
- Department of Vascular Surgery, Peking University People's Hospital, Peking University, Beijing, China (W.L.)
| | - Zihan Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| | - Yiting Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| | - Fang Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| | - Jian Luo
- Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China (J.L.)
| | - Yi Fu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| | - Jinpeng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, China (R.Z., J.S.)
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China (Y.C., C.M., R.G., Z.M., Y.J., F.Y., Y.F., J.S., W.K.).,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China (Y.C., R.G., Z.M., Y.J., F.Y., Y.F., W.K.)
| |
Collapse
|
42
|
Barrasa-Ramos S, Dessalles CA, Hautefeuille M, Barakat AI. Mechanical regulation of the early stages of angiogenesis. J R Soc Interface 2022; 19:20220360. [PMID: 36475392 PMCID: PMC9727679 DOI: 10.1098/rsif.2022.0360] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Favouring or thwarting the development of a vascular network is essential in fields as diverse as oncology, cardiovascular disease or tissue engineering. As a result, understanding and controlling angiogenesis has become a major scientific challenge. Mechanical factors play a fundamental role in angiogenesis and can potentially be exploited for optimizing the architecture of the resulting vascular network. Largely focusing on in vitro systems but also supported by some in vivo evidence, the aim of this Highlight Review is dual. First, we describe the current knowledge with particular focus on the effects of fluid and solid mechanical stimuli on the early stages of the angiogenic process, most notably the destabilization of existing vessels and the initiation and elongation of new vessels. Second, we explore inherent difficulties in the field and propose future perspectives on the use of in vitro and physics-based modelling to overcome these difficulties.
Collapse
Affiliation(s)
- Sara Barrasa-Ramos
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Claire A. Dessalles
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| | - Mathieu Hautefeuille
- Laboratoire de Biologie du Développement (UMR7622), Institut de Biologie Paris Seine, Sorbonne Université, Paris, France,Facultad de Ciencias, Universidad Nacional Autónoma de México, CDMX, Mexico
| | - Abdul I. Barakat
- LadHyX, CNRS, Ecole Polytechnique, Institut Polytechnique de Paris, Palaiseau, France
| |
Collapse
|
43
|
Barbacena P, Dominguez-Cejudo M, Fonseca CG, Gómez-González M, Faure LM, Zarkada G, Pena A, Pezzarossa A, Ramalho D, Giarratano Y, Ouarné M, Barata D, Fortunato IC, Misikova LH, Mauldin I, Carvalho Y, Trepat X, Roca-Cusachs P, Eichmann A, Bernabeu MO, Franco CA. Competition for endothelial cell polarity drives vascular morphogenesis in the mouse retina. Dev Cell 2022; 57:2321-2333.e9. [PMID: 36220082 PMCID: PMC9552591 DOI: 10.1016/j.devcel.2022.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 07/15/2022] [Accepted: 09/07/2022] [Indexed: 12/02/2022]
Abstract
Blood-vessel formation generates unique vascular patterns in each individual. The principles governing the apparent stochasticity of this process remain to be elucidated. Using mathematical methods, we find that the transition between two fundamental vascular morphogenetic programs-sprouting angiogenesis and vascular remodeling-is established by a shift of collective front-to-rear polarity of endothelial cells in the mouse retina. We demonstrate that the competition between biochemical (VEGFA) and mechanical (blood-flow-induced shear stress) cues controls this collective polarity shift. Shear stress increases tension at focal adhesions overriding VEGFA-driven collective polarization, which relies on tension at adherens junctions. We propose that vascular morphogenetic cues compete to regulate individual cell polarity and migration through tension shifts that translates into tissue-level emergent behaviors, ultimately leading to uniquely organized vascular patterns.
Collapse
Affiliation(s)
- Pedro Barbacena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Dominguez-Cejudo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Catarina G Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Manuel Gómez-González
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Laura M Faure
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain
| | - Georgia Zarkada
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Andreia Pena
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Anna Pezzarossa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Champalimaud Foundation, Champalimaud Research, Lisbon, Portugal
| | - Daniela Ramalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ylenia Giarratano
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK
| | - Marie Ouarné
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - David Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Isabela C Fortunato
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Lenka Henao Misikova
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Ian Mauldin
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK; School of Informatics, The University of Edinburgh, Edinburgh, UK
| | - Yulia Carvalho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain; Facultat de Medicina, Universitat de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), the Barcelona Institute of Technology (BIST), Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA; Université de Paris, PARCC, INSERM, 75006 Paris, France
| | - Miguel O Bernabeu
- Centre for Medical Informatics, Usher Institute, The University of Edinburgh, Edinburgh, UK; The Bayes Centre, The University of Edinburgh, Edinburgh, UK
| | - Cláudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Universidade Católica Portuguesa, Católica Medical School, Católica Biomedical Research Centre, Lisbon, Portugal.
| |
Collapse
|
44
|
Zhang Z, Lai G, Sun L. Basement-Membrane-Related Gene Signature Predicts Prognosis in WHO Grade II/III Gliomas. Genes (Basel) 2022; 13:1810. [PMID: 36292695 PMCID: PMC9602375 DOI: 10.3390/genes13101810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/29/2022] [Accepted: 10/05/2022] [Indexed: 10/17/2023] Open
Abstract
Gliomas that are classified as grade II or grade III lesions by the World Health Organization (WHO) are highly aggressive, and some may develop into glioblastomas within a short period, thus portending the conferral of a poor prognosis for patients. Previous studies have implicated basement membrane (BM)-related genes in glioma development. In this study, we constructed a prognostic model for WHO grade II/III gliomas in accordance with the risk scores of BM-related genes. Differentially expressed genes (DEGs) in the glioma samples relative to normal samples were screened from the GEO database, and five prognostically relevant BM-related genes, including NELL2, UNC5A, TNC, CSPG4, and SMOC1, were selected using Cox regression analyses for the risk score model. The median risk score was calculated, based on which high- and low-risk groups of patients were generated. The clinical information, pathological information, and risk group were combined to establish a prognostic nomogram. Both the nomogram and risk score model performed well in the independent CGGA cohort. Gene set enrichment analysis (GSEA) and immune profile, drug sensitivity, and tumor mutation burden (TMB) analyses were performed in the two risk groups. A significant enrichment of 'Autophagy-other', 'Collecting duct acid secretion', 'Glycosphingolipid biosynthesis-lacto and neolacto series', 'Valine, leucine, and isoleucine degradation', 'Vibrio cholerae infection', and other pathways were observed for patients with high risk. In addition, higher proportions of monocytes and resting CD4 memory T cells were observed in the low- and high-risk groups, respectively. In conclusion, the BM-related gene risk score model can guide the clinical management of WHO grade II and III gliomas.
Collapse
Affiliation(s)
- Zhaogang Zhang
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Guichuan Lai
- Department of Epidemiology and Health Statistics, School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Lingling Sun
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| |
Collapse
|
45
|
Chighizola M, Dini T, Marcotti S, D'Urso M, Piazzoni C, Borghi F, Previdi A, Ceriani L, Folliero C, Stramer B, Lenardi C, Milani P, Podestà A, Schulte C. The glycocalyx affects the mechanotransductive perception of the topographical microenvironment. J Nanobiotechnology 2022; 20:418. [PMID: 36123687 PMCID: PMC9484177 DOI: 10.1186/s12951-022-01585-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
The cell/microenvironment interface is the starting point of integrin-mediated mechanotransduction, but many details of mechanotransductive signal integration remain elusive due to the complexity of the involved (extra)cellular structures, such as the glycocalyx. We used nano-bio-interfaces reproducing the complex nanotopographical features of the extracellular matrix to analyse the glycocalyx impact on PC12 cell mechanosensing at the nanoscale (e.g., by force spectroscopy with functionalised probes). Our data demonstrates that the glycocalyx configuration affects spatio-temporal nanotopography-sensitive mechanotransductive events at the cell/microenvironment interface. Opposing effects of major glycocalyx removal were observed, when comparing flat and specific nanotopographical conditions. The excessive retrograde actin flow speed and force loading are strongly reduced on certain nanotopographies upon strong reduction of the native glycocalyx, while on the flat substrate we observe the opposite trend. Our results highlight the importance of the glycocalyx configuration in a molecular clutch force loading-dependent cellular mechanism for mechanosensing of microenvironmental nanotopographical features.
Collapse
Affiliation(s)
- Matteo Chighizola
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Tania Dini
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Stefania Marcotti
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Mirko D'Urso
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,Department of Biomedical Engineering, Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Claudio Piazzoni
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Francesca Borghi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Anita Previdi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Laura Ceriani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Claudia Folliero
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.,The FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Brian Stramer
- Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Cristina Lenardi
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Paolo Milani
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy
| | - Alessandro Podestà
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| | - Carsten Schulte
- Interdisciplinary Centre for Nanostructured Materials and Interfaces (C.I.Ma.I.Na.) and Department of Physics "Aldo Pontremoli", University of Milan, Milan, Italy.
| |
Collapse
|
46
|
Youn J, Kim DS. Engineering porous membranes mimicking in vivo basement membrane for organ-on-chips applications. BIOMICROFLUIDICS 2022; 16:051301. [PMID: 36275917 PMCID: PMC9586704 DOI: 10.1063/5.0101397] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/16/2022] [Indexed: 06/16/2023]
Abstract
Porous membrane-based microfluidic chips are frequently used for developing in vitro tissue-barrier models, the so-called tissue barriers-on-chips (TBoCs). The porous membrane in a TBoC plays a crucial role as an alternative to an in vivo basement membrane (BM). To improve the physiological relevance of an artificial porous membrane, it should possess complex BM-like characteristics from both biophysical and biochemical perspectives. For practical use, artificial membranes should have high mechanical robustness, and their fabrication processes should be conducive to mass production. There have been numerous approaches to accomplishing these requirements in BM-like porous membranes. Extracellular matrix (ECM) hydrogels have emerged as physiologically relevant materials for developing artificial BMs; they remarkably improve the phenotypes and functions of both cells and their layers when compared to previous synthetic porous membranes. However, for practical use, the poor mechanical robustness of ECM membranes needs to be improved. Recently, an advanced ECM membrane reinforced with a nanofiber scaffold has been introduced that possesses both BM-like characteristics and practical applicability. This advanced ECM membrane is expected to promote not only in vivo-like cellular functions but also cellular responses to drugs, which in turn further facilitates the practical applications of TBoCs.
Collapse
Affiliation(s)
- Jaeseung Youn
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | | |
Collapse
|
47
|
Caliandro MF, Schmalbein F, Todesca LM, Mörgelin M, Rezaei M, Meißner J, Siepe I, Grosche J, Schwab A, Eble JA. A redox-dependent thiol-switch and a Ca 2+ binding site within the hinge region hierarchically depend on each other in α7β1 integrin regulation. Free Radic Biol Med 2022; 187:38-49. [PMID: 35605898 DOI: 10.1016/j.freeradbiomed.2022.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/17/2022] [Indexed: 11/23/2022]
Abstract
Integrin-mediated cell contacts with the extracellular matrix (ECM) are essential for cellular adhesion, force transmission, and migration. Several effectors, such as divalent cations and redox-active compounds, regulate ligand binding activities of integrins and influence their cellular functions. To study the role of the Ca2+ binding site within the hinge region of the integrin α7 subunit, we genetically abrogated it in the α7hiΔCa mutant. This mutant folded correctly, associated with the β1 subunit and was exposed on the cell surface, but showed reduced ligand binding and weaker cell adhesion to laminin-111. Thus, it resembles the α7hiΔSS mutant, in which the redox-regulated pair of cysteines, closeby to the Ca2+ binding site within the hinge, was abrogated. Comparing both mutants in adhesion strength and cell migration revealed that both Ca2+ complexation and redox-regulation within the hinge interdepend on each other. Moreover, protein-chemical analyses of soluble integrin ectodomains containing the same α7 hinge mutations suggest that integrin activation via the subunit α hinge is primed by the formation of the cysteine pair-based crosslinkage. Then, this allows Ca2+ complexation within the hinge, which is another essential step for integrin activation and ligand binding. Thus, the α hinge is an allosteric integrin regulation site, in which both effectors, Ca2+ and redox-active compounds, synergistically and hierarchically induce far-ranging conformational changes, such as the extension of the integrin ectodomain, resulting in integrin activation of ECM ligand binding and altered integrin-mediated cell functions.
Collapse
Affiliation(s)
- Michele F Caliandro
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Felix Schmalbein
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Luca Matteo Todesca
- University of Münster, Institute of Physiology II, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | | | - Maryam Rezaei
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Juliane Meißner
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Isabel Siepe
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Julius Grosche
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Johannes A Eble
- University of Münster, Institute of Physiological Chemistry and Pathobiochemistry, Waldeyerstr. 15, 48149, Münster, Germany.
| |
Collapse
|
48
|
Mancinelli E, Takuma M, Fujie T, Pensabene V. Recreating cellular barriers in human microphysiological systems in-vitro. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:3923-3926. [PMID: 36086504 DOI: 10.1109/embc48229.2022.9870981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Within cellular barriers, cells are separated by basement membranes (BMs), nanometer-thick extracellular matrix layers. In existing in-vitro cellular-barrier models, cell-to-cell signaling can be preserved by culturing different cells in individual chambers separated by a semipermeable membrane. Their structure does not always replicate the BM thickness nor diffusion through it. Here, a porous polymeric nanofilm made of poly(D-L-lactic acid) (PDLLA) is proposed to recreate the BM in a microfluidic blood-brain-barrier model. Nanofilms showed an average thickness of [Formula: see text] and a maximum pore diameter of 1.6 μm. Human umbilical vein endothelial cells (HUVECs) were cultured on PDLLA. After 7 days, viability was >95% and cell morphology did not show relevant differences with HUVECs grown on control substrates. A protocol for suspending the nanofilm between 2 microfluidic chambers was identified and showed no leakage and good sealing. Clinical Relevance- Preclinical models of cellular barriers are a key step towards a deeper understanding of their roles in pathogenesis of various diseases: a physiologically relevant microfluidic model of the blood brain barrier (BBB) allows high-throughput investigations of BBB contribution in neurodegenerative diseases and cruelty-free screenings of drugs targeting the brain.
Collapse
|
49
|
Topography-induced large-scale antiparallel collective migration in vascular endothelium. Nat Commun 2022; 13:2797. [PMID: 35589751 PMCID: PMC9120158 DOI: 10.1038/s41467-022-30488-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Collective migration of vascular endothelial cells is central for embryonic development, angiogenesis, and wound closure. Although physical confinement of cell assemblies has been shown to elicit specific patterns of collective movement in various cell types, endothelial migration in vivo often occurs without confinement. Here we show that unconfined endothelial cell monolayers on microgroove substrates that mimic the anisotropic organization of the extracellular matrix exhibit a specific type of collective movement that takes the form of a periodic pattern of antiparallel cell streams. We further establish that the development of these streams requires intact cell-cell junctions and that stream sizes are particularly sensitive to groove depth. Finally, we show that modeling the endothelial cell sheet as an active fluid with the microgrooves acting as constraints on cell orientation predicts the occurrence of the periodic antiparallel cell streams as well as their lengths and widths. We posit that in unconfined cell assemblies, physical factors that constrain or bias cellular orientation such as anisotropic extracellular matrix cues or directed flow-derived shear forces dictate the pattern of collective cell movement. The physical environment dictates the emergence of specific patterns of collective cell migration. Here, authors show that unconfined endothelial monolayers on microgroove substrates exhibit an original pattern of antiparallel cell streams.
Collapse
|
50
|
|